Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 371(3): 633-641, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31578258

RESUMO

N-methyl-d-aspartate (NMDA) receptor-dependent long-term potentiation (LTP) is an established cellular model underlying learning and memory, and involves intracellular signaling mediated by the second messenger cyclic guanosine monophosphate (cGMP). As phosphodiesterase (PDE)9A selectively hydrolyses cGMP in areas of the brain related to cognition, PDE9A inhibitors may improve cognitive function by enhancing NMDA receptor-dependent LTP. This study aimed to pharmacologically characterize BI 409306, a novel PDE9A inhibitor, using in vitro assays and in vivo determination of cGMP levels in the brain. Further, the effects of BI 409306 on synaptic plasticity evaluated by LTP in ex vivo hippocampal slices and on cognitive performance in rodents were also investigated. In vitro assays demonstrated that BI 409306 is a potent and selective inhibitor of human and rat PDE9A with mean concentrations at half-maximal inhibition (IC50) of 65 and 168 nM. BI 409306 increased cGMP levels in rat prefrontal cortex and cerebrospinal fluid and attenuated a reduction in mouse striatum cGMP induced by the NMDA-receptor antagonist MK-801. In ex vivo rat brain slices, BI 409306 enhanced LTP induced by both weak and strong tetanic stimulation. Treatment of mice with BI 409306 reversed MK-801-induced working memory deficits in a T-maze spontaneous-alternation task and improved long-term memory in an object recognition task. These findings suggest that BI 409306 is a potent and selective inhibitor of PDE9A. BI 409306 shows target engagement by increasing cGMP levels in brain, facilitates synaptic plasticity as demonstrated by enhancement of hippocampal LTP, and improves episodic and working memory function in rodents. SIGNIFICANCE STATEMENT: This preclinical study demonstrates that BI 409306 is a potent and selective PDE9A inhibitor in rodents. Treatment with BI 409306 increased brain cGMP levels, promoted long-term potentiation, and improved episodic and working memory performance in rodents. These findings support a role for PDE9A in synaptic plasticity and cognition. The potential benefits of BI 409306 are currently being investigated in clinical trials.


Assuntos
Química Encefálica/efeitos dos fármacos , GMP Cíclico/análise , Memória/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Pirazóis/farmacocinética , Pirimidinas/farmacocinética , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , Animais , GMP Cíclico/líquido cefalorraquidiano , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Wistar
2.
EMBO J ; 33(17): 1912-27, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25024434

RESUMO

Neuronal histone acetylation has been linked to memory consolidation, and targeting histone acetylation has emerged as a promising therapeutic strategy for neuropsychiatric diseases. However, the role of histone-modifying enzymes in the adult brain is still far from being understood. Here we use RNA sequencing to screen the levels of all known histone acetyltransferases (HATs) in the hippocampal CA1 region and find that K-acetyltransferase 2a (Kat2a)--a HAT that has not been studied for its role in memory function so far--shows highest expression. Mice that lack Kat2a show impaired hippocampal synaptic plasticity and long-term memory consolidation. We furthermore show that Kat2a regulates a highly interconnected hippocampal gene expression network linked to neuroactive receptor signaling via a mechanism that involves nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). In conclusion, our data establish Kat2a as a novel and essential regulator of hippocampal memory consolidation.


Assuntos
Regulação da Expressão Gênica , Redes Reguladoras de Genes , Histona Acetiltransferases/metabolismo , Memória , Animais , Região CA1 Hipocampal/enzimologia , Perfilação da Expressão Gênica , Histona Acetiltransferases/genética , Camundongos , Camundongos Knockout
3.
Nature ; 485(7400): 651-5, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22660329

RESUMO

Extracellular plaques of amyloid-ß and intraneuronal neurofibrillary tangles made from tau are the histopathological signatures of Alzheimer's disease. Plaques comprise amyloid-ß fibrils that assemble from monomeric and oligomeric intermediates, and are prognostic indicators of Alzheimer's disease. Despite the importance of plaques to Alzheimer's disease, oligomers are considered to be the principal toxic forms of amyloid-ß. Interestingly, many adverse responses to amyloid-ß, such as cytotoxicity, microtubule loss, impaired memory and learning, and neuritic degeneration, are greatly amplified by tau expression. Amino-terminally truncated, pyroglutamylated (pE) forms of amyloid-ß are strongly associated with Alzheimer's disease, are more toxic than amyloid-ß, residues 1-42 (Aß(1-42)) and Aß(1-40), and have been proposed as initiators of Alzheimer's disease pathogenesis. Here we report a mechanism by which pE-Aß may trigger Alzheimer's disease. Aß(3(pE)-42) co-oligomerizes with excess Aß(1-42) to form metastable low-n oligomers (LNOs) that are structurally distinct and far more cytotoxic to cultured neurons than comparable LNOs made from Aß(1-42) alone. Tau is required for cytotoxicity, and LNOs comprising 5% Aß(3(pE)-42) plus 95% Aß(1-42) (5% pE-Aß) seed new cytotoxic LNOs through multiple serial dilutions into Aß(1-42) monomers in the absence of additional Aß(3(pE)-42). LNOs isolated from human Alzheimer's disease brain contained Aß(3(pE)-42), and enhanced Aß(3(pE)-42) formation in mice triggered neuron loss and gliosis at 3 months, but not in a tau-null background. We conclude that Aß(3(pE)-42) confers tau-dependent neuronal death and causes template-induced misfolding of Aß(1-42) into structurally distinct LNOs that propagate by a prion-like mechanism. Our results raise the possibility that Aß(3(pE)-42) acts similarly at a primary step in Alzheimer's disease pathogenesis.


Assuntos
Peptídeos beta-Amiloides/química , Amiloide/toxicidade , Ácido Glutâmico/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/toxicidade , Fragmentos de Peptídeos/química , Príons/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Amiloide/química , Amiloide/efeitos dos fármacos , Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Modelos Animais de Doenças , Ácido Glutâmico/química , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Príons/química , Príons/toxicidade , Proteínas tau/deficiência , Proteínas tau/genética
4.
Proc Natl Acad Sci U S A ; 110(2): 648-53, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23267072

RESUMO

The coagulation protease activated protein C (aPC) confers cytoprotective effects in various in vitro and in vivo disease models, including diabetic nephropathy. The nephroprotective effect may be related to antioxidant effects of aPC. However, the mechanism through which aPC may convey these antioxidant effects and the functional relevance of these properties remain unknown. Here, we show that endogenous and exogenous aPC prevents glomerular accumulation of oxidative stress markers and of the redox-regulating protein p66(Shc) in experimental diabetic nephropathy. These effects were predominately observed in podocytes. In vitro, aPC inhibited glucose-induced expression of p66(Shc) mRNA and protein in podocytes (via PAR-1 and PAR-3) and various endothelial cell lines, but not in glomerular endothelial cells. Treatment with aPC reversed glucose-induced hypomethylation and hyperacetylation of the p66(Shc) promoter in podocytes. The hyperacetylating agent sodium butyrate abolished the suppressive effect of aPC on p66(Shc) expression both in vitro and in vivo. Moreover, sodium butyrate abolished the beneficial effects of aPC in experimental diabetic nephropathy. Inhibition of p66(Shc) expression and mitochondrial translocation by aPC normalized mitochondrial ROS production and the mitochondrial membrane potential in glucose-treated podocytes. Genetic ablation of p66(Shc) compensated for the loss of protein C activation in vivo, normalizing markers of diabetic nephropathy and oxidative stress. These studies identify a unique mechanism underlying the cytoprotective effect of aPC. Activated PC epigenetically controls expression of the redox-regulating protein p66(Shc), thus linking the extracellular protease aPC to mitochondrial function in diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/tratamento farmacológico , Repressão Epigenética/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Proteína C/farmacologia , Proteínas Adaptadoras da Sinalização Shc/antagonistas & inibidores , Análise de Variância , Animais , Butiratos/farmacologia , Imunoprecipitação da Cromatina , Metilação de DNA/efeitos dos fármacos , Primers do DNA/genética , Nefropatias Diabéticas/etiologia , Técnicas de Silenciamento de Genes , Immunoblotting , Imuno-Histoquímica , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/metabolismo , Podócitos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Estatísticas não Paramétricas , Frações Subcelulares
5.
Neural Plast ; 2016: 4145708, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26881108

RESUMO

The neurotrophin brain derived neurotrophic factor (BDNF) is an important growth factor in the CNS. Deficits in transport of this secretory protein could underlie neurodegenerative diseases. Investigation of disease-related changes in BDNF transport might provide insights into the cellular mechanism underlying, for example, Alzheimer's disease (AD). To analyze the role of BDNF transport in AD, live cell imaging of fluorescently labeled BDNF was performed in hippocampal neurons of different AD model systems. BDNF and APP colocalized with low incidence in vesicular structures. Anterograde as well as retrograde transport of BDNF vesicles was reduced and these effects were mediated by factors released from hippocampal neurons into the extracellular medium. Transport of BDNF was altered at a very early time point after onset of human APP expression or after acute amyloid-beta(1-42) treatment, while the activity-dependent release of BDNF remained unaffected. Taken together, extracellular cleavage products of APP induced rapid changes in anterograde and retrograde transport of BDNF-containing vesicles while release of BDNF was unaffected by transgenic expression of mutated APP. These early transport deficits might lead to permanently impaired brain functions in the adult brain.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Fragmentos de Peptídeos/toxicidade , Vesículas Transportadoras/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Hipocampo/efeitos dos fármacos , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Vesículas Transportadoras/efeitos dos fármacos
6.
Kidney Int ; 87(1): 74-84, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25075770

RESUMO

Diabetic nephropathy is a growing health concern with characteristic sterile inflammation. As the underlying mechanisms of this inflammation remain poorly defined, specific therapies targeting sterile inflammation in diabetic nephropathy are lacking. Intriguingly, an association of diabetic nephropathy with inflammasome activation has recently been shown, but the pathophysiological relevance of this finding remains unknown. Within glomeruli, inflammasome activation was detected in endothelial cells and podocytes in diabetic humans and mice and in glucose-stressed glomerular endothelial cells and podocytes in vitro. Abolishing Nlrp3 or caspase-1 expression in bone marrow-derived cells fails to protect mice against diabetic nephropathy. Conversely, Nlrp3-deficient mice are protected against diabetic nephropathy despite transplantation of wild-type bone marrow. Pharmacological IL-1R antagonism prevented or even reversed diabetic nephropathy in mice. Mitochondrial reactive oxygen species (ROS) activate the Nlrp3 inflammasome in glucose or advanced glycation end product stressed podocytes. Inhibition of mitochondrial ROS prevents glomerular inflammasome activation and nephropathy in diabetic mice. Thus, mitochondrial ROS and Nlrp3-inflammasome activation in non-myeloid-derived cells aggravate diabetic nephropathy. Targeting the inflammasome may be a potential therapeutic approach to diabetic nephropathy.


Assuntos
Proteínas de Transporte/imunologia , Nefropatias Diabéticas/imunologia , Inflamassomos/imunologia , Glomérulos Renais/citologia , Animais , Células Endoteliais/imunologia , Humanos , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Podócitos/imunologia , Índice de Gravidade de Doença
7.
Acta Neuropathol ; 129(2): 259-77, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25391494

RESUMO

Neuronal injury from ischemic stroke is aggravated by invading peripheral immune cells. Early infiltrates of neutrophil granulocytes and T-cells influence the outcome of stroke. So far, however, neither the timing nor the cellular dynamics of neutrophil entry, its consequences for the invaded brain area, or the relative importance of T-cells has been extensively studied in an intravital setting. Here, we have used intravital two-photon microscopy to document neutrophils and brain-resident microglia in mice after induction of experimental stroke. We demonstrated that neutrophils immediately rolled, firmly adhered, and transmigrated at sites of endothelial activation in stroke-affected brain areas. The ensuing neutrophil invasion was associated with local blood-brain barrier breakdown and infarct formation. Brain-resident microglia recognized both endothelial damage and neutrophil invasion. In a cooperative manner, they formed cytoplasmic processes to physically shield activated endothelia and trap infiltrating neutrophils. Interestingly, the systemic blockade of very-late-antigen-4 immediately and very effectively inhibited the endothelial interaction and brain entry of neutrophils. This treatment thereby strongly reduced the ischemic tissue injury and effectively protected the mice from stroke-associated behavioral impairment. Behavioral preservation was also equally well achieved with the antibody-mediated depletion of myeloid cells or specifically neutrophils. In contrast, T-cell depletion more effectively reduced the infarct volume without improving the behavioral performance. Thus, neutrophil invasion of the ischemic brain is rapid, massive, and a key mediator of functional impairment, while peripheral T-cells promote brain damage. Acutely depleting T-cells and inhibiting brain infiltration of neutrophils might, therefore, be a powerful early stroke treatment.


Assuntos
Isquemia Encefálica/imunologia , Integrina alfa4beta1/metabolismo , Microglia/fisiologia , Infiltração de Neutrófilos/fisiologia , Neutrófilos/fisiologia , Acidente Vascular Cerebral/imunologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Encéfalo/imunologia , Encéfalo/patologia , Isquemia Encefálica/patologia , Adesão Celular/fisiologia , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Atividade Motora/fisiologia , Neutrófilos/patologia , Distribuição Aleatória , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/patologia
8.
Proc Natl Acad Sci U S A ; 109(31): 12503-8, 2012 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-22814377

RESUMO

Oligomers are intermediates of the ß-amyloid (Aß) peptide fibrillogenic pathway and are putative pathogenic culprits in Alzheimer's disease (AD). Here we report the biotechnological generation and biochemical characterization of an oligomer-specific antibody fragment, KW1. KW1 not only discriminates between oligomers and other Aß conformations, such as fibrils or disaggregated peptide; it also differentiates between different types of Aß oligomers, such as those formed by Aß (1-40) and Aß (1-42) peptide. This high selectivity of binding contrasts sharply with many other conformational antibodies that interact with a large number of structurally analogous but sequentially different antigens. X-ray crystallography, NMR spectroscopy, and peptide array measurements imply that KW1 recognizes oligomers through a hydrophobic and significantly aromatic surface motif that includes Aß residues 18-20. KW1-positive oligomers occur in human AD brain samples and induce synaptic dysfunctions in living brain tissues. Bivalent KW1 potently neutralizes this effect and interferes with Aß assembly. By altering a specific step of the fibrillogenic cascade, it prevents the formation of mature Aß fibrils and induces the accumulation of nonfibrillar aggregates. Our data illuminate significant mechanistic differences in oligomeric and fibril recognition and suggest the considerable potential of KW1 in future studies to detect or inhibit specific types of Aß conformers.


Assuntos
Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/química , Multimerização Proteica , Motivos de Aminoácidos , Anticorpos Monoclonais , Cristalografia por Raios X , Humanos , Ressonância Magnética Nuclear Biomolecular , Estrutura Quaternária de Proteína
9.
Neuroimage ; 103: 171-180, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25234116

RESUMO

Electrical and optogenetic methods for brain stimulation are widely used in rodents for manipulating behavior and analyzing functional connectivities in neuronal circuits. High-resolution in vivo imaging of the global, brain-wide, activation patterns induced by these stimulations has remained challenging, in particular in awake behaving mice. We here mapped brain activation patterns in awake, intracranially self-stimulating mice using a novel protocol for single-photon emission computed tomography (SPECT) imaging of regional cerebral blood flow (rCBF). Mice were implanted with either electrodes for electrical stimulation of the medial forebrain bundle (mfb-microstim) or with optical fibers for blue-light stimulation of channelrhodopsin-2 expressing neurons in the ventral tegmental area (vta-optostim). After training for self-stimulation by current or light application, respectively, mice were implanted with jugular vein catheters and intravenously injected with the flow tracer 99m-technetium hexamethylpropyleneamine oxime (99mTc-HMPAO) during seven to ten minutes of intracranial self-stimulation or ongoing behavior without stimulation. The 99mTc-brain distributions were mapped in anesthetized animals after stimulation using multipinhole SPECT. Upon self-stimulation rCBF strongly increased at the electrode tip in mfb-microstim mice. In vta-optostim mice peak activations were found outside the stimulation site. Partly overlapping brain-wide networks of activations and deactivations were found in both groups. When testing all self-stimulating mice against all controls highly significant activations were found in the rostromedial nucleus accumbens shell. SPECT-imaging of rCBF using intravenous tracer-injection during ongoing behavior is a new tool for imaging regional brain activation patterns in awake behaving rodents providing higher spatial and temporal resolutions than 18F-2-fluoro-2-dexoyglucose positron emission tomography.


Assuntos
Mapeamento Encefálico/métodos , Encéfalo/diagnóstico por imagem , Circulação Cerebrovascular/fisiologia , Optogenética/métodos , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Encéfalo/irrigação sanguínea , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Compostos Radiofarmacêuticos , Recompensa , Autoestimulação , Tecnécio Tc 99m Exametazima
10.
Proc Natl Acad Sci U S A ; 107(5): 1942-7, 2010 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-20133839

RESUMO

The formation of extracellular amyloid plaques is a common patho-biochemical event underlying several debilitating human conditions, including Alzheimer's disease (AD). Considerable evidence implies that AD damage arises primarily from small oligomeric amyloid forms of Abeta peptide, but the precise mechanism of pathogenicity remains to be established. Using a cell culture system that reproducibly leads to the formation of Alzheimer's Abeta amyloid plaques, we show here that the formation of a single amyloid plaque represents a template-dependent process that critically involves the presence of endocytosis- or phagocytosis-competent cells. Internalized Abeta peptide becomes sorted to multivesicular bodies where fibrils grow out, thus penetrating the vesicular membrane. Upon plaque formation, cells undergo cell death and intracellular amyloid structures become released into the extracellular space. These data imply a mechanism where the pathogenic activity of Abeta is attributed, at least in part, to intracellular aggregates.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/ultraestrutura , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Técnica de Fratura por Congelamento , Humanos , Líquido Intracelular/metabolismo , Camundongos , Microscopia Eletrônica de Varredura , Microscopia de Vídeo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Placa Amiloide/química , Placa Amiloide/ultraestrutura
11.
J Neurosci ; 31(36): 12790-801, 2011 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-21900558

RESUMO

Posttranslational amyloid-ß (Aß) modification is considered to play an important role in Alzheimer's disease (AD) etiology. An N-terminally modified Aß species, pyroglutamate-amyloid-ß (pE3-Aß), has been described as a major constituent of Aß deposits specific to human AD but absent in normal aging. Formed via cyclization of truncated Aß species by glutaminyl cyclase (QC; QPCT) and/or its isoenzyme (isoQC; QPCTL), pE3-Aß aggregates rapidly and is known to seed additional Aß aggregation. To directly investigate pE3-Aß toxicity in vivo, we generated and characterized transgenic TBA2.1 and TBA2.2 mice, which express truncated mutant human Aß. Along with a rapidly developing behavioral phenotype, these mice showed progressively accumulating Aß and pE3-Aß deposits in brain regions of neuronal loss, impaired long-term potentiation, microglial activation, and astrocytosis. Illustrating a threshold for pE3-Aß neurotoxicity, this phenotype was not found in heterozygous animals but in homozygous TBA2.1 or double-heterozygous TBA2.1/2.2 animals only. A significant amount of pE3-Aß formation was shown to be QC-dependent, because crossbreeding of TBA2.1 with QC knock-out, but not isoQC knock-out, mice significantly reduced pE3-Aß levels. Hence, lowering the rate of QC-dependent posttranslational pE3-Aß formation can, in turn, lower the amount of neurotoxic Aß species in AD.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Transtornos Heredodegenerativos do Sistema Nervoso/patologia , Hipocampo/patologia , Ácido Pirrolidonocarboxílico/metabolismo , Envelhecimento/patologia , Envelhecimento/psicologia , Doença de Alzheimer/patologia , Animais , Comportamento Animal , Encéfalo/patologia , Ensaio de Imunoadsorção Enzimática , Gliose/patologia , Transtornos Heredodegenerativos do Sistema Nervoso/psicologia , Humanos , Imuno-Histoquímica , Cinética , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Microscopia Eletrônica , Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Fenótipo , Equilíbrio Postural/fisiologia , Processamento de Proteína Pós-Traducional , Reflexo de Sobressalto/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
J Neurochem ; 121(5): 774-84, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22375951

RESUMO

Pyroglutamate (pGlu)-modified amyloid peptides have been identified in sporadic and familial forms of Alzheimer's disease (AD) and the inherited disorders familial British and Danish Dementia (FBD and FDD). In this study, we characterized the aggregation of amyloid-ß protein Aß37, Aß38, Aß40, Aß42 and ADan species in vitro, which were modified by N-terminal pGlu (pGlu-Aß3-x, pGlu-ADan) or possess the intact N-terminus (Aß1-x, ADan). The pGlu-modification confers rapid formation of oligomers and short fibrillar aggregates. In accordance with these observations, the pGlu-modified Aß38, Αß40 and Αß42 species inhibit hippocampal long term potentiation of synaptic response, but pGlu-Aß3-42 showing the highest effect. Among the unmodified Aß peptides, only Aß1-42 exhibites such propensity, which was similar to pGlu-Aß3-38 and pGlu-Aß3-40. Likewise, the amyloidogenic peptide pGlu-ADan impaired synaptic potentiation more pronounced than N-terminal unmodified ADan. The results were validated using conditioned media from cultivated HEK293 cells, which express APP variants favoring the formation of Aß1-x, Aß3-x or N-truncated pGlu-Aß3-x species. Hence, we show that the ability of different amyloid peptides to impair synaptic function apparently correlates to their potential to form oligomers as a common mechanism. The pGlu-modification is apparently mediating a higher surface hydrophobicity, as shown by 1-anilinonaphtalene-8-sulfonate fluorescence, which enforces potential to interfere with neuronal physiology.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Potenciação de Longa Duração/fisiologia , Ácido Pirrolidonocarboxílico/metabolismo , Peptídeos beta-Amiloides/química , Animais , Eletroforese em Gel de Poliacrilamida , Células HEK293 , Hipocampo/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ácido Pirrolidonocarboxílico/química
13.
J Neurosci Res ; 90(1): 193-202, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21971686

RESUMO

Reactive oxygen species (ROS) are key players in ischemia-induced neurodegeneration. We investigated whether hippocampal neurons may lack sufficient redox-buffering capacity to protect against ROS attacks. Using organotypic hippocampal slice cultures (OHSCs) transiently exposed to oxygen and glucose deprivation (OGD) and gerbils suffering from a two-vessel occlusion (2VO) as complementary ex vivo and in vivo models, we have elucidated whether the intrinsic redox systems interfere with ischemia-induced neurodegeneration. Cell- type-specific immunohistological staining of hippocampal slice cultures revealed that pyramidal neurons, in contrast to astrocytes and microglia, express free thiols only weakly. In addition, free thiol levels were extensively decreased throughout the hippocampal formation immediately after OGD, but recovered within 24 hr after reperfusion. In parallel, progressive glia activation and proliferation were observed. Increased neuronal exposure to ROS was monitored by dihydroethidium oxidation in hippocampal pyramidal cell layers immediately after OGD. Coadministration of reduction equivalents (α-lipoic acid) and thiol-stimulating agents (enalapril, ambroxol) decreased ischemia-induced neuronal damage in OGD-treated OHSCs and in gerbils exposed to 2VO, whereas single drug applications remained ineffective. In summary, limited redox buffering capacities of pyramidal neurons may underlie their exceptional vulnerability to cerebral ischemia. Consistently, multidrug treatments supporting endogenous redox systems may offer a strategy to promote valid neuroprotection.


Assuntos
Isquemia Encefálica/patologia , Neurônios/patologia , Oxirredução , Traumatismo por Reperfusão/patologia , Animais , Morte Celular , Modelos Animais de Doenças , Etídio/análogos & derivados , Etídio/metabolismo , Fluoresceínas/metabolismo , Gerbillinae , Proteína Glial Fibrilar Ácida/metabolismo , Glucose/deficiência , Glicoproteínas/metabolismo , Hipocampo/citologia , Hipóxia , Lectinas/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio , Rodaminas/metabolismo , Compostos de Sulfidrila/metabolismo , Ácido Tióctico/farmacologia , Versicanas
15.
Exp Cell Res ; 315(1): 50-66, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18992240

RESUMO

The bone marrow represents an easy accessible source of adult stem cells suitable for various cell based therapies. Several studies in recent years suggested the existence of pluripotent stem cells within bone marrow stem cells (BMSC) expressing marker proteins of both embryonic and tissue committed stem cells. These subpopulations were referred to as MAPC, MIAMI and VSEL-cells. Here we describe SD-BMSC (serumdeprivation-induced BMSC) which are induced as a distinct subpopulation after complete serumdeprivation. SD-BMSC are generated from small-sized nestin-positive BMSC (S-BMSC) organized as round-shaped cells in the top layer of BMSC-cultures. The generation of SD-BMSC is caused by a selective proliferation of S-BMSC and accompanied by changes in both morphology and gene expression. SD-BMSC up-regulate not only markers typical for neural stem cells like nestin and GFAP, but also proteins characteristic for embryonic cells like Oct4 and SOX2. We hypothesize, that SD-BMSC like MAPC, MIAMI and VSEL-cells represent derivatives from a single pluripotent stem cell fraction within BMSC exhibiting characteristics of embryonic and tissue committed stem cells. The complete removal of serum might offer a simple way to specifically enrich this fraction of pluripotent embryonic like stem cells in BMSC cultures.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Embrionárias/citologia , Neurônios/citologia , Soro/metabolismo , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/metabolismo , Polaridade Celular , Proliferação de Células , Forma Celular , Células Cultivadas , Estruturas Celulares/metabolismo , Fenômenos Eletrofisiológicos , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Proteínas de Filamentos Intermediários/metabolismo , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Ratos , Ratos Sprague-Dawley , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo
16.
J Neurosci ; 28(23): 5965-75, 2008 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-18524901

RESUMO

Microglial cells maintain the immunological integrity of the healthy brain and can exert protection from traumatic injury. During ischemic tissue damage such as stroke, peripheral immune cells acutely infiltrate the brain and may exacerbate neurodegeneration. Whether and how microglia can protect from this insult is unknown. Polymorphonuclear neutrophils (PMNs) are a prominent immunologic infiltrate of ischemic lesions in vivo. Here, we show in organotypic brain slices that externally applied invading PMNs massively enhance ischemic neurotoxicity. This, however, is counteracted by additional application of microglia. Time-lapse imaging shows that microglia exert protection by rapid engulfment of apoptotic, but, strikingly, also viable, motile PMNs in cell culture and within brain slices. PMN engulfment is mediated by integrin- and lectin-based recognition. Interference with this process using RGDS peptides and N-acetyl-glucosamine blocks engulfment of PMNs and completely abrogates the neuroprotective function of microglia. Thus, engulfment of invading PMNs by microglia may represent an entirely new mechanism of CNS immune privilege.


Assuntos
Movimento Celular/imunologia , Sistema Nervoso Central/imunologia , Microglia/imunologia , Neurônios/imunologia , Neutrófilos/imunologia , Fagocitose/imunologia , Animais , Células Cultivadas , Sistema Nervoso Central/citologia , Imunidade Celular , Camundongos , Neurônios/citologia , Neutrófilos/patologia , Ratos , Ratos Wistar
17.
J Neuroimmunol ; 206(1-2): 91-9, 2009 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-19028400

RESUMO

Inflammatory reactions in the CNS, resulting from a loss of control and involving a network of non-neuronal and neuronal cells, are major contributors to the onset and progress of several major neurodegenerative diseases. Therapeutic strategies should therefore keep or restore the well-controlled and finely-tuned balance of immune reactions, and protect neurons from inflammatory damage. In our study, we selected plants of the Malaysian rain forest by an ethnobotanic survey, and investigated them in cell-based-assay-systems and in living brain tissue cultures in order to identify anti-inflammatory and neuroprotective effects. We found that alcoholic extracts from the tropical plant Knema laurina (Black wild nutmeg) exhibited highly anti-inflammatory and neuroprotective effects in cell culture experiments, reduced NO- and IL-6-release from activated microglia cells dose-dependently, and protected living brain tissue from microglia-mediated inflammatory damage at a concentration of 30 microg/ml. On the intracellular level, the extract inhibited ERK-1/2-phosphorylation, IkB-phosphorylation and subsequently NF-kB-translocation in microglia cells. K. laurina belongs to the family of Myristicaceae, which have been used for centuries for treatment of digestive and inflammatory diseases and is also a major food plant of the Giant Hornbill. Moreover, extract from K. laurina promotes also neurogenesis in living brain tissue after oxygen-glucose deprivation. In conclusion, extract from K. laurina not only controls and limits inflammatory reaction after primary neuronal damage, it promotes moreover neurogenesis if given hours until days after stroke-like injury.


Assuntos
Encefalite/tratamento farmacológico , Myristicaceae/química , Neurogênese/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Fitoterapia , Extratos Vegetais/uso terapêutico , Análise de Variância , Animais , Animais Recém-Nascidos , Bromodesoxiuridina/metabolismo , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteínas do Domínio Duplacortina , Encefalite/etiologia , Glucose/deficiência , Ácido Glutâmico/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipóxia/tratamento farmacológico , Infarto da Artéria Cerebral Média/complicações , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Microglia/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Técnicas de Cultura de Órgãos , Extratos Vegetais/química
18.
J Neurosci Methods ; 177(1): 149-59, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19007815

RESUMO

Bromodeoxyuridine incorporates into DNA during mitosis. A long-term stability of the incorporated BrdU is important for the recovery of BrdU-labeled cells. For testing the stability of BrdU incorporation into DNA we pulse-labeled mesenchymal stem cells with BrdU and observed these cells in vitro over 4 weeks. During this time the BrdU-signal was permanently decreasing. Starting with cells containing evenly stained BrdU-nuclei, so-called filled cells, already 3 days after BrdU removal we detected cells containing so-called segmented and punctated BrdU-signals. The number of those labeled cells continuously increased over time. Interestingly, the loss of BrdU in the nucleus was accompanied by an increasing labeling of the cytosol. Further, we injected BrdU intraperitoneally into rats after ischemia and detected BrdU-positive cells in the hippocampus 3 and 23 days after the last BrdU injection. While after 3 days most of the BrdU-positive cells in the hippocampus displayed a filled BrdU-signal, 23 days after BrdU removal an increased number of segmented and punctated BrdU-positive nuclei was detected. The gradual degradation of the BrdU-signal was not caused by cell death. The consequence of this BrdU degradation would be an underestimation of cell proliferation and an overestimation of cell death of newly generated cells.


Assuntos
Células da Medula Óssea/citologia , Bromodesoxiuridina/metabolismo , Proliferação de Células , Células-Tronco Mesenquimais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Caspase 3/metabolismo , Contagem de Células , Células Cultivadas , Citocromos c/metabolismo , Proteínas do Domínio Duplacortina , Inibidores Enzimáticos/farmacologia , Hipocampo/patologia , Isquemia/patologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas , Estaurosporina/farmacologia , Fatores de Tempo
19.
J Pharmacol Exp Ther ; 327(2): 343-52, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18701765

RESUMO

N-Methyl-D-aspartate (NMDA) receptor-mediated excitotoxicity is thought to underlie a variety of neurological disorders, and inhibition of either the NMDA receptor itself, or molecules of the intracellular cascade, may attenuate neurodegeneration in these diseases. Calpain, a calcium-dependent cysteine protease, has been identified as part of such an NMDA receptor-induced excitotoxic signaling pathway. The present study addressed the question of whether inhibition of calpain can prevent neuronal cell death and associated behavioral deficits in a disease-relevant animal model, which is based on excitotoxic lesions of the cholinergic nucleus basalis magnocellularis of Meynert. Excitotoxic lesions of the nucleus basalis with NMDA induced a markedly impaired performance in the novel object recognition test. Treatment with the calpain inhibitor, N-(1-benzyl-2-carbamoyl-2-oxoethyl)-2-[E-2-(4-diethlyaminomethylphenyl) ethen-1-yl]benzamide (A-705253), dose-dependently prevented the behavioral deficit. Subsequent analysis of choline acetyltransferase in the cortical mantle of the lesioned animals revealed that application of A-705253 dose-dependently and significantly attenuated cholinergic neurodegeneration. Calpain inhibition also significantly diminished the accompanying gliosis, as determined by immunohistochemical analysis of microglia activation. Finally, inhibition of calpain by A-705253 and the peptidic calpain inhibitor N-acetyl-Leu-Leu-Nle-CHO did not impair long-term potentiation in hippocampal slices, indicating that calpain inhibition interrupts NMDA excitotoxicity pathways without interfering with NMDA receptor-mediated signaling involved in cognition. We conclude that inhibition of calpains may represent a valuable strategy for the prevention of excitotoxicity-induced neuronal decline without interfering with the physiological neuronal functions associated with learning and memory processes. Thus, calpain inhibition may be a promising and novel approach for the treatment of various neurodegenerative disorders.


Assuntos
Núcleo Basal de Meynert/efeitos dos fármacos , Benzamidas/farmacologia , Calpaína/antagonistas & inibidores , N-Metilaspartato/toxicidade , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Animais , Núcleo Basal de Meynert/patologia , Cognição/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Microglia/efeitos dos fármacos , Microglia/fisiologia , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/fisiologia
20.
Neuropharmacology ; 55(5): 908-18, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18674549

RESUMO

The present study investigated the putative pro-cognitive effects of the novel selective PDE9 inhibitor BAY 73-6691. The effects on basal synaptic transmission and long-term potentiation (LTP) were investigated in rat hippocampal slices. Pro-cognitive effects were assessed in a series of learning and memory tasks using rodents as subjects. BAY 73-6691 had no effect on basal synaptic transmission in hippocampal slices prepared from young adult (7- to 8-week-old) Wistar rats. A dose of 10 microM, but not 30 microM, BAY 73-6691 enhanced early LTP after weak tetanic stimulation. The dose effective in young adult Wistar rats did not affect LTP in hippocampal slices prepared from young (7- to 8-week-old) Fischer 344 X Brown Norway (FBNF1) rats, probably reflecting strain differences. However, it increased basal synaptic transmission and enhanced early LTP after weak tetanic stimulation in hippocampal slices prepared from very old (31- to 35-month-old) FBNF1 rats. BAY 73-6691 enhanced acquisition, consolidation, and retention of long-term memory (LTM) in a social recognition task and tended to enhance LTM in an object recognition task. Bay 73-6691 attenuated the scoplamine-induced retention deficit in a passive avoidance task, and the MK-801-induced short-term memory deficits in a T-maze alternation task. The mechanism of action, possibly through modulation of the NO/cGMP-PKG/CREB pathway, is discussed. Our findings support the notion that PDE9 inhibition may be a novel target for treating memory deficits that are associated with aging and neurodegenerative disorders such as Alzheimer's disease.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Reconhecimento Visual de Modelos/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento de Escolha/efeitos dos fármacos , Antagonistas Colinérgicos/farmacologia , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Estimulação Elétrica , Inibidores Enzimáticos/química , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Hipocampo/efeitos da radiação , Técnicas In Vitro , Potenciação de Longa Duração/fisiologia , Potenciação de Longa Duração/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pirazóis/química , Pirimidinas/química , Ratos , Ratos Wistar , Tempo de Reação/efeitos dos fármacos , Escopolamina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA